Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Pharmacol Res Perspect ; 12(4): e1226, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38886975

RESUMEN

Although classically recognized as a neurotransmitter, gamma aminobutyric acid (GABA) has also been identified in colonic tumors. Moreover, the gut microbiome represents another potential source of GABA. Both GABAA and GABAB receptors have been implicated in contributing to the effects of GABA in colorectal cancer, with both pro- and anti-tumorigenic functions identified. However, their subunit composition is often overlooked. Studies to date have not addressed whether the GABA-producing potential of the microbiome changes over the course of colon tumor development or whether receptor subunit expression patterns are altered in colon cancer. Therefore, we investigated the clusters of orthologous group frequencies of glutamate decarboxylase (GAD) in feces from two murine models of colon cancer and found that the frequency of microbial GAD was significantly decreased early in the tumorigenic process. We also determined that microbial-derived GABA inhibited proliferation of colon cancer cells in vitro and that this effect of GABA on SW480 cells involved both GABAA and GABAB receptors. GABA also inhibited prostaglandin E2 (PGE2)-induced proliferation and interleukin-6 (IL-6) expression in these cells. Gene expression correlations were assessed using the "Cancer Exploration" suite of the TIMER2.0 web tool and identified that GABA receptor subunits were differentially expressed in human colon cancer. Moreover, GABAA receptor subunits were predominantly positively associated with PGE2 synthase, cyclooxygenase-2 and IL-6. Collectively, these data demonstrate decreased potential of the microbiome to produce GABA during tumorigenesis, a novel anti-tumorigenic pathway for GABA, and that GABA receptor subunit expression adds a further layer of complexity to GABAergic signaling in colon cancer.


Asunto(s)
Proliferación Celular , Neoplasias del Colon , Microbioma Gastrointestinal , Receptores de GABA-A , Receptores de GABA-B , Transducción de Señal , Ácido gamma-Aminobutírico , Animales , Neoplasias del Colon/metabolismo , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Ácido gamma-Aminobutírico/metabolismo , Humanos , Ratones , Línea Celular Tumoral , Receptores de GABA-A/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-B/metabolismo , Dinoprostona/metabolismo , Glutamato Descarboxilasa/metabolismo , Interleucina-6/metabolismo , Ciclooxigenasa 2/metabolismo , Ciclooxigenasa 2/genética , Carcinogénesis , Heces/microbiología , Receptores de GABA/metabolismo , Receptores de GABA/genética , Masculino , Ratones Endogámicos C57BL , Femenino
2.
Int J Food Microbiol ; 317: 108385, 2020 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-31783343

RESUMEN

Due to its ubiquitous nature, Listeria monocytogenes is a threat to all fresh fruits and vegetables, including mushrooms, which are Ireland's largest horticultural crop. Although fresh cultivated mushrooms (Agaricus bisporus) have not been previously linked with listeriosis outbreaks, the pathogen still poses a threat to the industry, particularly due to its ability to form biofilms. This threat is highlighted by the multiple recalls of mushroom products caused by L. monocytogenes contamination and by previous studies demonstrating that L. monocytogenes is present in the mushroom production environment. In this study, the biofilm formation potential of L. monocytogenes strains isolated from the mushroom production environment was investigated on materials and at temperatures relevant to mushroom production. A preliminary assessment of biofilm formation of 73 mushroom industry isolates was undertaken using a crystal violet assay on polystyrene microtitre plates. The biofilm formation of a subset (n = 7) of these strains was then assessed on twelve different materials, including materials that are representative of the materials commonly found in the mushroom production environments, using the CDC biofilm reactor. Vertical scanning interferometry was used to determine the surface roughness of the chosen materials. All the strains tested using the CDC biofilm reactor were able to form biofilms on the different surfaces tested but material type was found to be a key determining factor on the levels of biofilm formed. Stainless steel, aluminium, rubber, polypropylene and polycarbonate were all able to support biofilm levels in the range of 4-4.9 log10 CFU/cm2, for seven different L. monocytogenes strains. Mushroom industry-specific materials, including growing nets and tarpaulins, were found to support biofilms levels between 4.7 and 6.7 log10 CFU/cm2. Concrete was found to be of concern as it supported 7.7 log10 CFU/cm2 of biofilm for the same strains; however, sealing the concrete resulted in an approximately 2-log reduction in biofilm levels. The surface roughness of the materials varied greatly between the materials (0.7-3.5 log10 Ra) and was found to have a positive correlation with biofilm formation (rs = 0.573) although marginally significant (P = 0.051). The results of this study indicate that L. monocytogenes can readily form biofilms on mushroom industry relevant surfaces, and additionally identifies surfaces of specific concern, where rigorous cleaning and disinfection is required.


Asunto(s)
Agaricales/fisiología , Biopelículas/crecimiento & desarrollo , Contaminación de Alimentos/prevención & control , Listeria monocytogenes/crecimiento & desarrollo , Aluminio , Desinfección/métodos , Microbiología de Alimentos , Irlanda , Cemento de Policarboxilato , Poliestirenos , Goma , Acero Inoxidable , Temperatura
3.
BMC Microbiol ; 19(1): 33, 2019 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-30736731

RESUMEN

BACKGROUND: Lactobacillus mucosae DPC 6426 has previously demonstrated potentially cardio-protective properties, in the form of dyslipidaemia and hypercholesterolemia correction in an apolipoprotein-E deficient mouse model. This study aims to characterise the manner in which this microbe may modulate host bile pool composition and immune response, in the context of cardiovascular disease. Lactobacillus mucosae DPC 6426 was assessed for bile salt hydrolase activity and specificity. The microbe was compared against several other enteric strains of the same species, as well as a confirmed bile salt hydrolase-active strain, Lactobacillus reuteri APC 2587. RESULTS: Quantitative bile salt hydrolase assays revealed that enzymatic extracts from Lactobacillus reuteri APC 2587 and Lactobacillus mucosae DPC 6426 demonstrate the greatest activity in vitro. Bile acid profiling of porcine and murine bile following incubation with Lactobacillus mucosae DPC 6426 confirmed a preference for hydrolysis of glyco-conjugated bile acids. In addition, the purified exopolysaccharide and secretome of Lactobacillus mucosae DPC 6426 were investigated for immunomodulatory capabilities using RAW264.7 macrophages. Gene expression data revealed that both fractions stimulated increases in interleukin-6 and interleukin-10 gene transcription in the murine macrophages, while the entire secretome was necessary to increase CD206 transcription. Moreover, the exopolysaccharide elicited a dose-dependent increase in nitric oxide and interleukin-10 production from RAW264.7 macrophages, concurrent with increased tumour necrosis factor-α secretion at all doses. CONCLUSIONS: This study indicates that Lactobacillus mucosae DPC 6426 modulates both bile pool composition and immune system tone in a manner which may contribute significantly to the previously identified cardio-protective phenotype.


Asunto(s)
Amidohidrolasas/biosíntesis , Bilis/metabolismo , Inmunomodulación , Lactobacillus/enzimología , Lactobacillus/inmunología , Macrófagos/inmunología , Animales , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/microbiología , Glicosiltransferasas/metabolismo , Hidrólisis , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Limosilactobacillus reuteri/enzimología , Lectinas Tipo C/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/microbiología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Óxido Nítrico/metabolismo , Polisacáridos Bacterianos/farmacología , Células RAW 264.7 , Receptores de Superficie Celular/metabolismo , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
4.
Mol Pharm ; 15(12): 5711-5727, 2018 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-30388019

RESUMEN

Pharmacokinetic research at the host-microbe interface has been primarily directed toward effects on drug metabolism, with fewer investigations considering the absorption process. We previously demonstrated that the transcriptional expression of genes encoding intestinal transporters involved in lipid translocation are altered in germ-free and conventionalized mice possessing distinct bile acid signatures. It was consequently hypothesized that microbial bile acid metabolism, which is the deconjugation and dehydroxylation of the bile acid steroid nucleus by gut bacteria, may impact upon drug transporter expression and/or activity and potentially alter drug disposition. Using a panel of three human intestinal cell lines (Caco-2, T84, and HT-29) that differ in basal transporter expression level, bile acid conjugation-, and hydroxylation-status was shown to influence the transcription of genes encoding several major influx and efflux transporter proteins. We further investigated if these effects on transporter mRNA would translate to altered drug disposition and activity. The results demonstrated that the conjugation and hydroxylation status of the bile acid steroid nucleus can influence the cellular response to multidrug resistance (MDR) substrates, a finding that did not directly correlate with directionality of gene or protein expression. In particular, we noted that the cytotoxicity of cyclosporine A was significantly augmented in the presence of the unconjugated bile acids deoxycholic acid (DCA) and chenodeoxycholic acid (CDCA) in P-gp positive cell lines, as compared to their taurine/glycine-conjugated counterparts, implicating P-gp in the molecular response. Overall this work identifies a novel mechanism by which gut microbial metabolites may influence drug accumulation and suggests a potential role for the microbial bile acid-deconjugating enzyme bile salt hydrolase (BSH) in ameliorating multidrug resistance through the generation of bile acid species with the capacity to access and inhibit P-gp ATPase. The physicochemical property of nonionization is suggested to underpin the preferential ability of unconjugated bile acids to attenuate the efflux of P-gp substrates and to sensitize tumorigenic cells to cytotoxic therapeutics in vitro. This work provides new impetus to investigate whether perturbation of the gut microbiota, and thereby the bile acid component of the intestinal metabolome, could alter drug pharmacokinetics in vivo. These findings may additionally contribute to the development of less toxic P-gp modulators, which could overcome MDR.


Asunto(s)
Ácido Quenodesoxicólico/metabolismo , Ciclosporina/farmacología , Ácido Desoxicólico/metabolismo , Microbioma Gastrointestinal/fisiología , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Variación Biológica Poblacional , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos/fisiología , Glicina , Células HT29 , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , ARN Mensajero/metabolismo , Taurina/metabolismo , Pruebas de Toxicidad
5.
J Crohns Colitis ; 12(2): 204-216, 2018 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-29373727

RESUMEN

BACKGROUND AND AIMS: Alterations in short chain fatty acid metabolism, particularly butyrate, have been reported in inflammatory bowel disease, but results have been conflicting because of small study numbers and failure to distinguish disease type, activity or other variables such as diet. We performed a comparative assessment of the capacity of the microbiota for butyrate synthesis, by quantifying butyryl-CoA:acetate CoA-transferase [BCoAT] gene content in stool from patients with Crohn's disease [CD; n = 71], ulcerative colitis [UC; n = 58] and controls [n = 75], and determined whether it was related to active vs inactive inflammation, microbial diversity, and composition and/or dietary habits. METHODS: BCoAT gene content was quantified by quantitative polymerase chain reaction [qPCR]. Disease activity was assessed clinically and faecal calprotectin concentration measured. Microbial composition was determined by sequencing 16S rRNA gene. Dietary data were collected using an established food frequency questionnaire. RESULTS: Reduced butyrate-synthetic capacity was found in patients with active and inactive CD [p < 0.001 and p < 0.01, respectively], but only in active UC [p < 0.05]. In CD, low BCoAT gene content was associated with ileal location, stenotic behaviour, increased inflammation, lower microbial diversity, greater microbiota compositional change, and decreased butyrogenic taxa. Reduced BCoAT gene content in patients with CD was linked with a different regimen characterised by lower dietary fibre. CONCLUSIONS: Reduced butyrate-synthetic capacity of the microbiota is more evident in CD than UC and may relate to reduced fibre intake. The results suggest that simple replacement of butyrate per se may be therapeutically inadequate, whereas manipulation of microbial synthesis, perhaps by dietary means, may be more appropriate.


Asunto(s)
Ácido Butírico/metabolismo , Clostridiales/aislamiento & purificación , Coenzima A Transferasas/genética , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , ADN Bacteriano/análisis , Microbioma Gastrointestinal/genética , Adulto , Estudios de Casos y Controles , Clostridiales/genética , Dieta , Fibras de la Dieta , Heces/química , Femenino , Frutas , Microbioma Gastrointestinal/fisiología , Humanos , Complejo de Antígeno L1 de Leucocito/análisis , Masculino , Persona de Mediana Edad , ARN Ribosómico 16S/análisis , Verduras
6.
Cell Mol Gastroenterol Hepatol ; 4(1): 65-74, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28560290

RESUMEN

BACKGROUND & AIMS: Options for the prevention of short-bowel syndrome-associated liver disease (SBS-ALDs) are limited and often ineffective. The farnesoid X receptor (FXR) is a newly emerging pharmaceutical target and FXR agonists have been shown to ameliorate cholestasis and metabolic disorders. The aim of this study was to assess the efficacy of obeticholic acid (OCA) treatment in preventing SBS-ALDs. METHODS: Piglets underwent 75% small-bowel resection (SBS) or sham surgery (sham) and were assigned to either a daily dose of OCA (2.4 mg/kg/day) or were untreated. Clinical measures included weight gain and stool studies. Histologic features were assessed. Ultraperformance liquid chromatography tandem mass spectrometry was used to determine bile acid composition in end point bile and portal serum samples. Gene expression of key FXR targets was assessed in intestinal and hepatic tissues via quantitative polymerase chain reaction. RESULTS: OCA-treated SBS piglets showed decreased stool fat and altered liver histology when compared with nontreated SBS piglets. OCA prevented SBS-associated taurine depletion, however, further analysis of bile and portal serum samples indicated that OCA did not prevent SBS-associated alterations in bile acid composition. The expression of FXR target genes involved in bile acid transport and synthesis increased within the liver of SBS piglets after OCA administration whereas, paradoxically, intestinal expression of FXR target genes were decreased by OCA administration. CONCLUSIONS: Administration of OCA in SBS reduced fat malabsorption and altered bile acid composition, but did not prevent the development of SBS-ALDs. We postulate that extensive small resection impacts the ability of the remnant intestine to respond to FXR activation.

7.
PLoS One ; 11(12): e0167319, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27907092

RESUMEN

Disruptions to circadian rhythm in mice and humans have been associated with an increased risk of obesity and metabolic syndrome. The gut microbiota is known to be essential for the maintenance of circadian rhythm in the host suggesting a role for microbe-host interactions in the regulation of the peripheral circadian clock. Previous work suggested a role for gut bacterial bile salt hydrolase (BSH) activity in the regulation of host circadian gene expression. Here we demonstrate that unconjugated bile acids, known to be generated through the BSH activity of the gut microbiota, are potentially chronobiological regulators of host circadian gene expression. We utilised a synchronised Caco-2 epithelial colorectal cell model and demonstrated that unconjugated bile acids, but not the equivalent tauro-conjugated bile salts, enhance the expression levels of genes involved in circadian rhythm. In addition oral administration of mice with unconjugated bile acids significantly altered expression levels of circadian clock genes in the ileum and colon as well as the liver with significant changes to expression of hepatic regulators of circadian rhythm (including Dbp) and associated genes (Per2, Per3 and Cry2). The data demonstrate a potential mechanism for microbe-host crosstalk that significantly impacts upon host circadian gene expression.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Relojes Circadianos/genética , Regulación de la Expresión Génica , Animales , Ácidos y Sales Biliares/farmacología , Proteínas CLOCK/genética , Línea Celular Tumoral , Células Epiteliales , Microbioma Gastrointestinal , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Ratones , Modelos Biológicos , Especificidad de Órganos/genética
8.
Bioengineered ; 7(2): 116-9, 2016 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-27212260

RESUMEN

We propose a mechanism of action for the betL* mutation which is based on DNA topology. Removing a single thymine residue from the betL σ(A) promoter's -10 and -35 spacer results in a 'twist'-mediated activation of transcription which accounts for the osmotolerance phenotype observed for strains expressing betL*.


Asunto(s)
Regulación Bacteriana de la Expresión Génica , Listeria monocytogenes/genética , Proteínas Bacterianas/genética , Betaína/metabolismo , Proteínas Portadoras/genética
9.
Annu Rev Food Sci Technol ; 7: 313-33, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26772409

RESUMEN

Bile acids have emerged as important signaling molecules in the host, as they interact either locally or systemically with specific cellular receptors, in particular the farnesoid X receptor (FXR) and TGR5. These signaling functions influence systemic lipid and cholesterol metabolism, energy metabolism, immune homeostasis, and intestinal electrolyte balance. Through defined enzymatic activities, the gut microbiota can significantly modify the signaling properties of bile acids and therefore can have an impact upon host health. Alterations to the gut microbiota that influence bile acid metabolism are associated with metabolic disease, obesity, diarrhea, inflammatory bowel disease (IBD), Clostridium difficile infection, colorectal cancer, and hepatocellular carcinoma. Here, we examine the regulation of this gut-microbiota-liver axis in the context of bile acid metabolism and indicate how this pathway represents an important target for the development of new nutraceutical (diet and/or probiotics) and targeted pharmaceutical interventions.


Asunto(s)
Ácidos y Sales Biliares/fisiología , Suplementos Dietéticos , Microbioma Gastrointestinal/fisiología , Tecnología Farmacéutica , Envejecimiento , Ácidos y Sales Biliares/metabolismo , Dieta , Enfermedad , Metabolismo Energético , Estado de Salud , Humanos , Hígado/metabolismo , Probióticos , Receptores Citoplasmáticos y Nucleares/fisiología , Transducción de Señal , Aumento de Peso
10.
Biomed Res Int ; 2015: 350526, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26543856

RESUMEN

The aim of this study was to assess the occurrence of L. ivanovii in foods and food processing environments in Ireland, to track persistence, and to characterize the disease causing potential of the isolated strains. A total of 2,006 samples (432 food samples and 1,574 environmental swabs) were collected between March 2013 and March 2014 from 48 food business operators (FBOs) belonging to different production sectors (dairy, fish, meat, and fresh-cut vegetable). Six of the forty-eight FBOs had samples positive for L. ivanovii on at least one sampling occasion. L. ivanovii was present in fifteen samples (fourteen environmental samples and one food sample). All but one of those positive samples derived from the dairy sector, where L. ivanovii prevalence was 1.7%. Six distinguishable pulsotypes were obtained by PFGE analysis, with one pulsotype being persistent in the environment of a dairy food business. Sequence analysis of the sigB gene showed that fourteen isolates belonged to L. ivanovii subsp. londoniensis, while only one isolate was L. ivanovii subsp. ivanovii. Cell invasion assays demonstrated that the majority of L. ivanovii strains were comparable to L. monocytogenes EGDe in their ability to invade CACO-2 epithelial cells whilst four isolates had significantly higher invasion efficiencies.


Asunto(s)
Contaminación de Alimentos/análisis , Microbiología de Alimentos , Listeria/aislamiento & purificación , Listeria/patogenicidad , Animales , Células CACO-2 , Productos Lácteos/microbiología , Electroforesis en Gel de Campo Pulsado , Monitoreo del Ambiente/métodos , Células Epiteliales/metabolismo , Manipulación de Alimentos , Industria de Alimentos , Humanos , Irlanda , Listeriosis , Carne/microbiología , Alimentos Marinos/microbiología , Verduras/microbiología , Virulencia
11.
PLoS One ; 9(11): e112649, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25386947

RESUMEN

The glutamate decarboxylase (GAD) system has been shown to be important for the survival of Listeria monocytogenes in low pH environments. The bacterium can use this faculty to maintain pH homeostasis under acidic conditions. The accepted model for the GAD system proposes that the antiport of glutamate into the bacterial cell in exchange for γ-aminobutyric acid (GABA) is coupled to an intracellular decarboxylation reaction of glutamate into GABA that consumes protons and therefore facilitates pH homeostasis. Most strains of L. monocytogenes possess three decarboxylase genes (gadD1, D2 & D3) and two antiporter genes (gadT1 & gadT2). Here, we confirm that the gadD3 encodes a glutamate decarboxylase dedicated to the intracellular GAD system (GADi), which produces GABA from cytoplasmic glutamate in the absence of antiport activity. We also compare the functionality of the GAD system between two commonly studied reference strains, EGD-e and 10403S with differences in terms of acid resistance. Through functional genomics we show that EGD-e is unable to export GABA and relies exclusively in the GADi system, which is driven primarily by GadD3 in this strain. In contrast 10403S relies upon GadD2 to maintain both an intracellular and extracellular GAD system (GADi/GADe). Through experiments with a murinised variant of EGD-e (EGDm) in mice, we found that the GAD system plays a significant role in the overall virulence of this strain. Double mutants lacking either gadD1D3 or gadD2D3 of the GAD system displayed reduced acid tolerance and were significantly affected in their ability to cause infection following oral inoculation. Since EGDm exploits GADi but not GADe the results indicate that the GADi system makes a contribution to virulence within the mouse. Furthermore, we also provide evidence that there might be a separate line of evolution in the GAD system between two commonly used reference strains.


Asunto(s)
Proteínas Bacterianas/metabolismo , Regulación Bacteriana de la Expresión Génica , Glutamato Descarboxilasa/metabolismo , Listeria monocytogenes/metabolismo , Listeria monocytogenes/patogenicidad , Animales , Proteínas Bacterianas/genética , Línea Celular/microbiología , Evolución Molecular , Femenino , Técnicas de Silenciamiento del Gen , Glutamato Descarboxilasa/genética , Humanos , Concentración de Iones de Hidrógeno , Listeria monocytogenes/genética , Listeriosis/microbiología , Macrófagos/microbiología , Ratones Endogámicos BALB C , Familia de Multigenes , Mutación , Ácido gamma-Aminobutírico/metabolismo
12.
Methods Mol Biol ; 1157: 263-74, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24792565

RESUMEN

During systemic infection by Listeria monocytogenes the host develops a robust T cell-mediated immune response against the major immunodominant antigens of the pathogen. The enzyme-linked immuno-spot (ELISPOT) test is an accurate and reproducible means of measuring the extent of this T cell response. Here we describe a detailed ELISPOT protocol for measuring an epitope-specific CD8+ T cell-mediated immune response in mice vaccinated with low doses of L. monocytogenes. The basic approach can be easily adapted for the analysis of other vaccination regimes and target epitopes.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Técnicas para Inmunoenzimas/métodos , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Animales , Antígenos Bacterianos/inmunología , Epítopos de Linfocito T/inmunología , Femenino , Ratones , Ratones Endogámicos BALB C , Vacunación
13.
Methods Mol Biol ; 1141: 201-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24567141

RESUMEN

Certain bacteria have emerged as biological gene vectors with natural tumor specificity, capable of specifically delivering genes or gene products to the tumor environment when intravenously (i.v.) administered to rodent models. Here, we describe procedures for studying this phenomenon in vitro and in vivo for both invasive and noninvasive bacteria suitable for exploitation as tumor-specific therapeutic delivery vehicles, due to their ability to replicate specifically within tumors and/or mediate bacterial-mediated transfer of plasmid DNA to mammalian cells (bactofection).


Asunto(s)
Bifidobacterium/genética , Escherichia coli/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Listeria monocytogenes/genética , Neoplasias/terapia , Plásmidos/metabolismo , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Recuento de Colonia Microbiana , Expresión Génica , Genes Reporteros , Vectores Genéticos , Humanos , Inyecciones Intralesiones , Inyecciones Intravenosas , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/patología
14.
PLoS One ; 7(2): e30928, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363518

RESUMEN

Regulation of iron homeostasis in many pathogens is principally mediated by the ferric uptake regulator, Fur. Since acquisition of iron from the host is essential for the intracellular pathogen Listeria monocytogenes, we predicted the existence of Fur-regulated systems that support infection. We examined the contribution of nine Fur-regulated loci to the pathogenicity of L. monocytogenes in a murine model of infection. While mutating the majority of the genes failed to affect virulence, three mutants exhibited a significantly compromised virulence potential. Most striking was the role of the membrane protein we designate FrvA (Fur regulated virulence factor A; encoded by frvA [lmo0641]), which is absolutely required for the systemic phase of infection in mice and also for virulence in an alternative infection model, the Wax Moth Galleria mellonella. Further analysis of the ΔfrvA mutant revealed poor growth in iron deficient media and inhibition of growth by micromolar concentrations of haem or haemoglobin, a phenotype which may contribute to the attenuated growth of this mutant during infection. Uptake studies indicated that the ΔfrvA mutant is unaffected in the uptake of ferric citrate but demonstrates a significant increase in uptake of haem and haemin. The data suggest a potential role for FrvA as a haem exporter that functions, at least in part, to protect the cell against the potential toxicity of free haem.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana/efectos de los fármacos , Hemo/toxicidad , Listeria monocytogenes/enzimología , Listeria monocytogenes/patogenicidad , Adenosina Trifosfatasas/genética , Animales , Proteínas Bacterianas/genética , Secuencia de Bases , Bioensayo , Biología Computacional , Farmacorresistencia Bacteriana/genética , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Genes Bacterianos/genética , Sitios Genéticos/genética , Homeostasis/efectos de los fármacos , Hierro/metabolismo , Listeria monocytogenes/efectos de los fármacos , Listeria monocytogenes/genética , Listeriosis/microbiología , Ratones , Datos de Secuencia Molecular , Mariposas Nocturnas/efectos de los fármacos , Mariposas Nocturnas/microbiología , Mutagénesis Insercional/efectos de los fármacos , Mutagénesis Insercional/genética , Mutación/genética , Plásmidos/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Virulencia/efectos de los fármacos , Virulencia/genética
15.
Curr Gene Ther ; 12(1): 12-9, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22263920

RESUMEN

Bacterial production of visible light is a natural phenomenon occurring in marine (Vibrio and Photobacterium) and terrestrial (Photorhabdus) species. The mechanism underpinning light production in these organisms is similar and involves the oxidation of an aldehyde substrate in a reaction catalysed by the bacterial luciferase enzyme. The genes encoding the luciferase and a fatty acid reductase complex which synthesizes the substrate are contained in a single operon (the lux operon). This provides a useful reporter system as cloning the operon into a recipient host bacterium will generate visible light without the requirement to add exogenous substrate. The light can be detected in vivo in the living animal using a sensitive detection system and is therefore ideally suited to bioluminescence imaging protocols. The system has therefore been widely used to track bacteria during infection or colonisation of the host. As bacteria are currently being examined as bactofection vectors for gene delivery, particularly to tumour tissue, the use of bioluminescence imaging offers a powerful means to investigate vector amplification in situ. The implications of this technology for bacterial localization, tumour targeting and gene transfer (bactofection) studies are discussed.


Asunto(s)
Aldehído Oxidorreductasas/genética , Genes Reporteros , Luciferasas/genética , Photobacterium/aislamiento & purificación , Photorhabdus/aislamiento & purificación , Vibrio/aislamiento & purificación , Aldehído Oxidorreductasas/metabolismo , Animales , Clonación Molecular , Vectores Genéticos , Luz , Luciferasas/metabolismo , Ratones , Operón/genética
16.
PLoS Pathog ; 7(5): e1002040, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21589907

RESUMEN

Human blood Vγ9/Vδ2 T cells, monocytes and neutrophils share a responsiveness toward inflammatory chemokines and are rapidly recruited to sites of infection. Studying their interaction in vitro and relating these findings to in vivo observations in patients may therefore provide crucial insight into inflammatory events. Our present data demonstrate that Vγ9/Vδ2 T cells provide potent survival signals resulting in neutrophil activation and the release of the neutrophil chemoattractant CXCL8 (IL-8). In turn, Vγ9/Vδ2 T cells readily respond to neutrophils harboring phagocytosed bacteria, as evidenced by expression of CD69, interferon (IFN)-γ and tumor necrosis factor (TNF)-α. This response is dependent on the ability of these bacteria to produce the microbial metabolite (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate (HMB-PP), requires cell-cell contact of Vγ9/Vδ2 T cells with accessory monocytes through lymphocyte function-associated antigen-1 (LFA-1), and results in a TNF-α dependent proliferation of Vγ9/Vδ2 T cells. The antibiotic fosmidomycin, which targets the HMB-PP biosynthesis pathway, not only has a direct antibacterial effect on most HMB-PP producing bacteria but also possesses rapid anti-inflammatory properties by inhibiting γδ T cell responses in vitro. Patients with acute peritoneal-dialysis (PD)-associated bacterial peritonitis--characterized by an excessive influx of neutrophils and monocytes into the peritoneal cavity--show a selective activation of local Vγ9/Vδ2 T cells by HMB-PP producing but not by HMB-PP deficient bacterial pathogens. The γδ T cell-driven perpetuation of inflammatory responses during acute peritonitis is associated with elevated peritoneal levels of γδ T cells and TNF-α and detrimental clinical outcomes in infections caused by HMB-PP positive microorganisms. Taken together, our findings indicate a direct link between invading pathogens, neutrophils, monocytes and microbe-responsive γδ T cells in early infection and suggest novel diagnostic and therapeutic approaches.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas/inmunología , Neutrófilos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Adulto , Células Presentadoras de Antígenos/metabolismo , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Infecciones Bacterianas/microbiología , Comunicación Celular/inmunología , Diferenciación Celular/inmunología , Supervivencia Celular/inmunología , Células Cultivadas , Difosfatos/metabolismo , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-8/inmunología , Interleucina-8/metabolismo , Activación de Linfocitos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Activación Neutrófila/inmunología , Neutrófilos/metabolismo , Peritonitis/inmunología , Peritonitis/microbiología , Fagocitosis/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/metabolismo , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
17.
Curr Gene Ther ; 10(1): 34-45, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20156189

RESUMEN

The food-grade bacterium Lactococcus lactis has been extensively investigated during the last two decades as a delivery vector for therapeutic proteins, DNA and vaccine antigens. The bacterium represents a safe, genetically tractable vector capable of producing heterologous therapeutic proteins at mucosal sites. Here we review recent work in which recombinant L. lactis strains have been exploited as agents to treat inflammatory bowel disease, allergy and cancer. We also describe the ability of L. lactis to deliver proteins with adjuvant potential, vaccines and DNA and discuss the therapeutic possibilities of this approach.


Asunto(s)
Vectores Genéticos/genética , Lactococcus lactis/genética , Lactococcus lactis/metabolismo , Proteínas Recombinantes/metabolismo , Animales , Sistemas de Liberación de Medicamentos , Humanos , Ratones , Proteínas Recombinantes/genética , Vacunas/genética , Vacunas/uso terapéutico
18.
Curr Gene Ther ; 10(1): 46-55, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20158470

RESUMEN

The intracellular pathogen Listeria monocytogenes represents a promising therapeutic vector for the delivery of DNA, RNA or protein to cancer cells or to prime immune responses against tumour-specific antigens. A number of biological properties make L. monocytogenes a promising platform for development as a vector for either gene therapy or as an anti-cancer vaccine vector. L. monocytogenes is particularly efficient in mediating internalization into host cells. Once inside cells, the bacterium produces specific virulence factors which lyse the vaculolar membrane and allow escape into the cytoplasm. Once in the cytosol, L. monocytogenes is capable of actin-based motility and cell-to-cell spread without an extracellular phase. The cytoplasmic location of L. monocytogenes is significant as this potentiates entry of antigens into the MHC Class I antigen processing pathway leading to priming of specific CD8(+) T cell responses. The cytoplasmic location is also beneficial for the delivery of DNA (bactofection) by L. monocytogenes whilst cell-to-cell spread may facilitate access of the vector to cells throughout the tumour. Several preclinical studies have demonstrated the ability of L. monocytogenes for intracellular gene or protein delivery in vitro and in vivo, and this vector has also displayed safety and efficacy in clinical trial. Here, we review the features of the L. monocytogenes host-pathogen interaction that make this bacterium such an attractive candidate with which to induce appropriate therapeutic responses. We focus primarily upon work that has led to attenuation of the pathogen, demonstrated DNA, RNA or protein delivery to tumour cells as well as research that shows the efficacy of L. monocytogenes as a vector for tumour-specific vaccine delivery.


Asunto(s)
Antineoplásicos/uso terapéutico , Sistemas de Liberación de Medicamentos , Listeria monocytogenes/fisiología , Neoplasias/tratamiento farmacológico , Animales , Vectores Genéticos , Humanos , Listeria monocytogenes/genética
19.
Hum Gene Ther ; 21(4): 405-16, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20105075

RESUMEN

Bacteria-mediated transfer of plasmid DNA to mammalian cells (bactofection) has been shown to have significant potential as an approach to express heterologous proteins in various cell types. This is achieved through entry of the entire bacterium into cells, followed by release of plasmid DNA. In a murine model, we show that Listeria monocytogenes can invade and spread in tumors, and establish the use of Listeria to deliver genes to tumors in vivo. A novel approach to vector lysis and release of plasmid DNA through antibiotic administration was developed. Ampicillin administration facilitated both plasmid transfer and safety control of vector. To further improve on the gene delivery system, we selected a Listeria monocytogenes derivative that is more sensitive to ampicillin, and less pathogenic than the wild-type strain. Incorporation of a eukaryotic-transcribed lysin cassette in the plasmid further increased bacterial lysis. Successful gene delivery of firefly luciferase to growing tumors in murine models and to patient breast tumor samples ex vivo was achieved. The model described encompasses a three-phase treatment regimen, involving (1) intratumoral administration of vector followed by a period of vector spread, (2) systemic ampicillin administration to induce vector lysis and plasmid transfer, and (3) systemic administration of combined moxifloxacin and ampicillin to eliminate systemic vector. For the first time, our results reveal the potential of Listeria monocytogenes for in vivo gene delivery.


Asunto(s)
Adenocarcinoma , Neoplasias de la Mama , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Listeria monocytogenes/genética , Plásmidos/genética , Adenocarcinoma/genética , Adenocarcinoma/microbiología , Adenocarcinoma/terapia , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/microbiología , Neoplasias de la Mama/terapia , Células CACO-2/microbiología , Línea Celular Tumoral , ADN Bacteriano/genética , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Listeria monocytogenes/patogenicidad , Listeriosis/microbiología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos
20.
Bioeng Bugs ; 1(4): 284-7, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-21327062

RESUMEN

Listeria monocytogenes is an intracellular pathogen that lyses the phagosomal vacuole of infected cells, proliferates in the host cell cytoplasm and can actively enter adjacent cells. The pathogen is therefore well suited to exploitation as a vector for the delivery of DNA to target cells as the lifecycle favors cellular targeting with vector amplification and the potential for cell-to-cell spread. We have recently demonstrated DNA transfer by L. monocytogenes in growing tumors in murine models. Our approach exploited an ampicillin sensitive stain of L. monocytogenes which can be lysed through systemic administration of ampicillin to facilitate release of plasmid DNA for expression by infected mammalian cells. Here, we discuss the implications of this technology and the potential for future improvements of the system.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/genética , Listeria monocytogenes/genética , Neoplasias/terapia , Animales , Línea Celular Tumoral , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA